Determination of the effects of lactoferrin in a preclinical mouse model of experimental colitis

  • Authors:
    • Li Li
    • Fazheng Ren
    • Zhanyou Yun
    • Ying An
    • Caiyun Wang
    • Xudong Yan
  • View Affiliations

  • Published online on: August 14, 2013     https://doi.org/10.3892/mmr.2013.1632
  • Pages: 1125-1129
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of this study was to determine the therapeutic efficacy of lactoferrin (Lf) on dextran sulphate sodium (DSS)‑induced experimental colitis in BALB/c mice. Eighty BALB/c mice were randomly divided into 4 groups; the normal, model, apo‑Lf and holo‑Lf groups. Fecal character, fecal occult blood, hematochezia and disease activity index (DAI) were recorded daily. The length of the colon was measured and histological scores were evaluated 28 days post‑treatment. Myeloperoxidase (MPO) activity was also determined and the expression of interleukin‑1β (IL‑1β) and tumor necrosis factor-α (TNF‑α) were measured by quantitative (q)PCR. Lf relieved the inflammatory condition of DSS‑induced experimental colitis in mice. The DAI and histological scores of Lf‑treated mice were lower compared with those of mice in the control group. The length of the colon of Lf‑treated mice was longer compared with that of mice in the control group. Treatment with Lf decreased MPO activity and the expression levels of IL‑1β and TNF‑α. In addition, Lf was found to promote beneficial effects in a mouse model of experimental colitis. Treatment with apo‑Lf was superior to that of holo‑Lf in the mouse model of DSS‑induced experimental colitis. Supplemental therapy with apo‑Lf may provide an important new tool in the clinical management of ulcerative colitis.

Introduction

Lactoferrin (Lf) is an 80 kDa, iron-binding glycoprotein, which is expressed abundantly in the exocrine secretions of mammals, particularly in milk and fluids of the digestive tract (1). Lf is also released by mucosal epithelia and neutrophils during inflammation (1).

Lf plays a direct antimicrobial role when present in mucosal membrane secretions of the epithelia and provides innate or mucosal immune defense by limiting the proliferation and adhesion of microbes and/or by microbicidal targeting. These properties are predominantly associated with the ability of Lf to sequester iron in biological fluids or in the destabilization of the membranes of microorganisms (2).

Iron-independent microbicidal activities, which require direct interaction between Lf and structural components of the surface of microbes, have also been demonstrated (1,2). In vitro and in vivo studies have shown that Lf may modulate adaptive and innate immune responses and thus protects the host against viral infections and other complex conditions, including septic shock (1,2).

Macrophages are important elements that provide innate immune defense against infection (3). During the course of an infection, macrophages are among the first cells in the major organs to be exposed to microbial challenges and are major producers of interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α) following infection (3).

Ulcerative colitis (UC) is a non-specific chronic inflammatory disease of the intestinal tract (4). Although the definitive causes of UC remain unclear, the major symptoms that characterize the disease include abdominal pain, diarrhea, presence of occult blood and mucus (5). Primary therapy of UC with various agents, including salazosulfamide, glucocorticoids and immunodepressant probiotics, has been attempted, but usually results in the poor treatment compliance of patients and increases rates of relapse of UC (6).

It was previously shown that gastrointestinal bacteria may be involved in the pathogenesis of UC (6). Thus, Lf has been increasingly considered as a therapeutic option in UC, due in part to Lf exhibiting multiple bioactivities. These bioactivites include restoration of the intestinal microbiota, anti-inflammatory properties and adjusting the intestinal immune response. Previously, Lf was observed to improve symptoms of UC in a rat model (7). In the current study, a model of DSS-induced colitis was used to study the therapeutic effect and mechanism of action of Lf in experimental UC.

Materials and methods

Lf

Lf was obtained from Tatua Co-operative Dairy Company, Limited (Tatua, New Zealand). To deplete iron, citric acid was added to the Lf solution and adjusted to pH 2.1 to obtain a final iron saturation of 7%. (NH4)2Fe(SO4)2 and HCO3 were added to the Lf solution to obtain a 100% iron-saturated Lf solution, as previously described (8).

Reagents

Dextran sulphate sodium (DSS) was provided as a 36–50 kDa reagent by MP Biomedicals (Santa Ana, CA, USA). All reagents required for quantitative (q)PCR analysis were obtained from Promega Corporation (Madison, WI, USA).

Experimental design

Equal numbers of male ten-week-old BALB/c mice, with a mean weight of 22.0±2.0 g, were obtained from Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). Mice were housed in clean filter-top cages under standard conditions of 50±10% humidity and an equal 12-h dark/light cycle and fed with standard mouse chow for 7 days. Mice were randomly divided into the following 4 groups: i) Normal, normal diet in the absence of any special treatment; ii) model, permitted to drink 2.5% DSS for modeling on days 15–21, plus normal saline (100 μl/10 g) by gavage on days 1–28; iii) apo-Lf, 2.5% DSS for modeling on days 15–21, plus apo-Lf (100 mg/kg/d) by gavage on days 1–28; and iv) holo-Lf, 2.5% DSS for modeling on days 15–21, plus holo-Lf (100 mg/kg/d) by gavage on days 1–28. On day 28, all mice were sacrificed.

To reflect the general conditions of the mice, disease activity index (DAI) scores were determined by an investigator blinded to the protocol. The extent of loss in body weight, fecal character, fecal occult blood or hematochezia was assessed as previously described (9). On day 28 following treatment, the colon was removed in the region that spanned the colo-cecal junction to the anus. The length of the resected tissue was measured, rinsed with 5 ml 0.01 mol/l PBS (pH 7.4) to remove fecal remnants and dissected longitudinally at the mesenteric attachment. Next, 5 mm of the distal colon was removed and fixed for 48 h in PBS buffered with 10% formalin. The tissue was then processed for paraffin embedding, cut into 5-μm sections and stained with hematoxylin and eosin. Other sections of the colon were preserved in liquid nitrogen. The study was approved by the Ethics Committee of the College of Veterinary Medicine, Inner Mongolia Agricultural University.

Disease activity

The following parameters were evaluated by statistical methods: Weight loss, fecal character, fecal occult blood and hematochezia (Table I) (9).

Table I

Disease activity index score.

Table I

Disease activity index score.

ScoreWeight loss (%)Fecal characterFecal bleeding
00NormalNone
11–5
2>5–10LooseHemoccult +
3>10–15
4>15WateryBleeding (visible)

[i] Normal feces, motions were well formed; loose, half-formed, pasty feces that did not adhere to the anus; watery, watery feces that adhered to the anus.

Histological scoring

Histological scores were given based on previously described criteria (10), with slight modifications. An extra score was added, which was denoted as 3.5 and represented observation of total crypt loss, but with evidence of retention to the surface epithelium. In addition, the lamina propria and sub-mucosa showed a serious inflammatory cellular infiltration in this group (Table II). Whole tissue specimens on slides were evaluated at magnification ×200 using light microscopy. Each microscopic field was evaluated and given a score. If more than one score was present in any given field of view, the scores were multiplied by the estimated percentage in the field and the sum of those values was calculated. At the conclusion of this evaluation process, the average score of the microscopic fields was recorded and analyzed statistically. The grading score was based on previously described criteria (10).

Table II

Histological scores for hematoxylin and eosin-stained colon sections.

Table II

Histological scores for hematoxylin and eosin-stained colon sections.

ScoreDescription
0.0Normal colonic mucosa
1.0Shortening and loss of the basal 1/3 of the crypts
2.0Loss of the basal 2/3 of the crypts
3.0Total crypt loss, but with retention to the epithelial surface
Lamina propria and sub-mucosa showed a mild inflammatory cellular infiltration
3.5Total crypt loss, but with retention to the epithelial surface
Lamina propria and submucosa showed a serious inflammatory cellular infiltration
4.0Erosions and marked cellular infiltration
Myeloperoxidase (MPO) activity

MPO activity was measured as an indicator of neutrophil accumulation in colonic mucosa as previously described (11).

qPCR

All primers (Table III) were designed using Primer Premier 5.0 software (Premier Biosoft, Palo Alto, CA, USA) and the primers were manufactured by Shinegene Molecular Biotechnology Co., Ltd. (Shanghai, China). The annealing temperatures ranged between 58 and 53°C for 45 min and amplification was performed using 30 cycles for each gene of interest.

Table III

PCR primers and products.

Table III

PCR primers and products.

Primer sequencesProduct size, bp
IL-1βF: AGCCCATCCTCTGTGACTCATG
R: GCTGATGTACCAGTTGGGGAAC
422
TNF-αF: GGCAGGTCTACTTTGGAGTCATTGC
R: CATTCGAGGCTCCAGTGAAATTCGG
299
GAPDHF: ACCACAGTCCATGCCATCAC
R: TCCACCACCCTGTTGCTGTA
440

[i] IL-1β, interleukin-1β; TNF-α, tumor necrosis factor-α.

Statistical analysis

Statistical analysis was performed using SPSS version 13.0 (SPSS, Inc., Chicago, IL, USA) software program for Windows. Data are presented as the mean ± SD. Data sets were analyzed by one-way analysis of variance (ANOVA) and Fisher’s protected LSD post-hoc test. Those values with a significant difference were further analyzed by the Student-Newman-Keuls test. P<0.05 was considered to indicate a statistically significant difference.

Results

Weight loss

Among the 4 groups of mice on day 1, no difference in body weight was observed (as determined by ANOVA). However, body weight increased gradually in the normal group. Mice in the model group showed weight loss over 1–14 days due to the gavage treatment received and a significant weight loss was observed from day 15 due to double stimulation by treatment with DSS and gavage. Treatment with holo-Lf and apo-Lf was observed to inhibit weight loss (Fig. 1).

Length of the colon

Length of the colon in the model group was significantly decreased compared with the normal group (P<0.05; Fig. 2). These observations indicate that Lf was capable of preventing shortening of the colon.

DAI scores

Weight loss, fecal character, fecal occult blood and hematochezia were evaluated individually as previously described (12). The highest DAI score was observed in the model group (Fig. 3). Lf also promoted a beneficial effect in experimental colitis. Low DAI scores were observed in the Lf groups (Fig. 3).

Histological scores

Histological changes in mice of the 4 groups were evaluated individually as previously described (10). The highest score was observed in mice of the model group and a lower score was observed in the Lf treatment groups when compared with the model group; this was particularly true in the context of mice in the apo-Lf treatment group (Fig. 4A and B).

MPO activity

Maximal MPO activity was observed to be 2.78±0.39 U/g in the model group, which was significantly higher than that noted in the control group (0.93±0.13 U/g) (P<0.05). MPO activities also decreased following Lf treatment (Fig. 5).

qPCR analysis

IL-1β mRNA levels were observed to be lowest in the normal group and at the highest levels in the model group (Fig. 6). In addition, levels of IL-1β mRNA were observed to be lower in the Lf treatment groups than in the model group (2.27±0.79 vs. 6.63±1.57; P<0.05). TNF-α mRNA levels were lowest in the normal group and highest in the model group (Fig. 6). In addition, TNF-α mRNA levels were observed to be lower in the Lf treatment group compared with the model group (1.73±0.46 vs. 5.23± 1.05; P<0.05; Fig. 6).

Discussion

UC is a form of intestinal inflammation, characterized by the diffuse infiltration of inflammatory cells and multiple ulcer formations in the colon. However, the causes and pathogenesis of UC remain unclear (13). In the current study, 2.5% DSS solution was administered to BALB/c mice in drinking water for seven days to establish a mouse model of DSS-induced colitis. This condition resembles human UC in the context of disease manifestations and pathological features (14).

Typical symptoms of colitis, including diarrhea, fecal occult blood and hematochezia, were observed in the colitis mice. In addition, intestinal histology reactions, including severe mucosal defects, hemorrhage, destruction of the glands and crypts, large and deep ulcer formation and massive inflammatory cell infiltrates were observed under light microscopy. Episodes of diarrhea, presence of fecal occult blood and hematochezia were observed to be markedly improved following oral treatment with Lf. Notably, intestinal mucosal inflammation and the shortening of the colon were significantly improved following oral Lf treatment. The DAI value and histology scores were significantly lower in the Lf treatment group when compared with the control group (P<0.05), indicating that Lf may therapeutically target and improve the symptoms of DSS-induced colitis.

A previous study showed that altered intestinal microenvironment and intestinal immunology are important in the pathogenesis of UC (15). Cytokines, including IL-1β and TNF-α, are not only important mediators of the immune response, but have been the focus of studies into the pathogenesis of UC (16). During the acute phase of DSS-induced colitis, the massive infiltrates of inflammatory cells secreted high levels of inflammatory cytokines that provoked intestinal mucosal injury and exacerbated the intestinal inflammatory reaction (12). The current study showed that mRNA levels of IL-1β and TNF-α in the intestinal mucosa of DSS-induced mice were significantly increased. These cytokines may be significant in the pathogenesis of UC and exacerbate mucosal inflammation and cellular apoptosis (17,18).

In the current study, mRNA levels of IL-1β and TNF-α in colonic tissues were quantified by qPCR, which showed that the two cytokines were elevated in the DSS-induced UC model. In addition, oral Lf treatment decreased mRNA levels of IL-1β and TNF-α, indicating that Lf may have an anti-inflammatory effect, thus preventing UC formation and ameliorating inflammation by inhibiting the synthesis of inflammatory cytokines. The current study also showed that apo-Lf improved anti-inflammatory effects when compared with holo-Lf. This indicated that the anti-inflammatory effects of Lf may be associated with the saturation state of iron in Lf, which is similar to its reported microbicidal effect (19,20).

LPS is produced by gram-negative bacteria and is one of the major factors leading to a marked and uncontrollable inflammatory response (21). The occurrence of colitis was hypothesized to be inhibited by apo-Lf through a mechanism dependent on restoration of homeostasis of intestinal bacterial flora (data not shown). Therefore, apo-Lf may reinforce the suppression of the inflammatory responses by inhibiting cytokine and LPS production. Further studies must be performed to investigate the effects of apo-Lf on the treatment of UC.

Acknowledgements

The authors thank Mr Zhang Ming (Key Laboratory of Functional Dairy, Beijing, China). This study was financed partly by the Inner Mongolia Yili Industrial Group Co., Ltd. (Hohhot, China).

References

1 

Legrand D, Elass E, Carpentier M and Mazurier J: Lactoferrin: a modulator of immune and inflammatory responses. Cell Mol Life Sci. 62:2549–2559. 2005. View Article : Google Scholar : PubMed/NCBI

2 

Valenti P and Antonini G: Lactoferrin: an important host defence against microbial and viral attack. Cell Mol Life Sci. 62:2576–2587. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Mogensen SC and Virelizier JL: The interferon-macrophage alliance. Interferon. 8:55–84. 1987.PubMed/NCBI

4 

Jiang XL and Cui HF: An analysis of 10218 ulcerative colitis cases in China. World J Gastroenterol. 8:158–161. 2002.PubMed/NCBI

5 

Carvalho R and Hyams JS: Diagnosis and management of inflammatory bowel disease in children. Semin Pediatr Surg. 16:164–171. 2007. View Article : Google Scholar

6 

Shanahan F: Crohn’s disease. Lancet. 359:62–69. 2002.

7 

Togawa J, Nagase H, Tanaka K, et al: Lactoferrin reduces colitis in rats via modulation of the immune system and correction of cytokine imbalance. Am J Physiol Gastrointest Liver Physiol. 283:G187–G195. 2002. View Article : Google Scholar

8 

Lu RR: Isolation, purification and bioactive functions of lactoferrin (unpublished PhD thesis). Jiangnan University; 2003, (In Chinese).

9 

Hamamoto N, Maemura K, Hirata I, Murano M, Sasaki S and Katsu K: Inhibition of dextran sulphate sodium (DSS)-induced colitis in mice by intracolonically administered antibodies against adhesion molecules (endothelial leucocyte adhesion molecule-1 (ELAM-1) or intercellular adhesion molecule-1 (ICAM-1)). Clin Exp Immunol. 117:462–468. 1999. View Article : Google Scholar

10 

Cooper HS, Murthy SN, Shah RS and Sedergran DJ: Clinicopathologic study of dextran sulfate sodium experimental murine colitis. Lab Invest. 69:238–249. 1993.PubMed/NCBI

11 

Fabia R, Ar’Rajab A, Johansson ML, et al: The effect of exogenous administration of Lactobacillus reuteri R2LC and oat fiber on acetic acid-induced colitis in the rat. Scand J Gastroenterol. 28:155–162. 1993.PubMed/NCBI

12 

Egger B, Bajaj-Elliott M, MacDonald TT, Inglin R, Eysselein VE and Büchler MW: Characterisation of acute murine dextran sodium sulphate colitis: cytokine profile and dose dependency. Digestion. 62:240–248. 2000. View Article : Google Scholar : PubMed/NCBI

13 

Zhang SZ, Zhao XH and Zhang DC: Cellular and molecular immunopathogenesis of ulcerative colitis. Cell Mol Immunol. 3:35–40. 2006.PubMed/NCBI

14 

Yan Y, Kolachala V, Dalmasso G, et al: Temporal and spatial analysis of clinical and molecular parameters in dextran sodium sulfate induced colitis. PLoS One. 4:e60732009. View Article : Google Scholar : PubMed/NCBI

15 

Beagley KW and Elson CO: Cells and cytokines in mucosal immunity and inflammation. Gastroenterol Clin North Am. 21:347–366. 1992.PubMed/NCBI

16 

Ishiguro Y: Mucosal proinflammatory cytokine production correlates with endoscopic activity of ulcerative colitis. J Gastroenterol. 34:66–74. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Wang QY, Chen CL, Wang JD, Lai ZS, Ma Q and Zhang YL: Expression of pro-inflammation cytokines and activation of nuclear factor kappab in the intestinal mucosa of mice with ulcerative colitis. Di Yi Jun Yi Da Xue Xue Bao. 23:1202–1205. 2003.(In Chinese).

18 

Rachmilewitz D, Karmeli F, Shteingart S, Lee J, Takabayashi K and Raz E: Immunostimulatory oligonucleotides inhibit colonic proinflammatory cytokine production in ulcerative colitis. Inflamm Bowel Dis. 12:339–345. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Kim WS, Tanaka T, Kumura H and Shimazaki K: Lactoferrin-binding proteins in Bifidobacterium bifidum. Biochem Cell Biol. 80:91–94. 2002. View Article : Google Scholar

20 

Tomita M, Wakabayashi H, Yamauchi K, Teraguchi S and Hayasawa H: Bovine lactoferrin and lactoferricin derived from milk: production and applications. Biochem Cell Biol. 80:109–112. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Cheng XY and Zhang LL: The relation of fulminant hepatitis with Endotoxin and its receptors and inflammatory cytokines. Jiangxi Medical Journal. 41:114–117. 2006.(In Chinese).

Related Articles

Journal Cover

October 2013
Volume 8 Issue 4

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Li L, Ren F, Yun Z, An Y, Wang C and Yan X: Determination of the effects of lactoferrin in a preclinical mouse model of experimental colitis. Mol Med Rep 8: 1125-1129, 2013
APA
Li, L., Ren, F., Yun, Z., An, Y., Wang, C., & Yan, X. (2013). Determination of the effects of lactoferrin in a preclinical mouse model of experimental colitis. Molecular Medicine Reports, 8, 1125-1129. https://doi.org/10.3892/mmr.2013.1632
MLA
Li, L., Ren, F., Yun, Z., An, Y., Wang, C., Yan, X."Determination of the effects of lactoferrin in a preclinical mouse model of experimental colitis". Molecular Medicine Reports 8.4 (2013): 1125-1129.
Chicago
Li, L., Ren, F., Yun, Z., An, Y., Wang, C., Yan, X."Determination of the effects of lactoferrin in a preclinical mouse model of experimental colitis". Molecular Medicine Reports 8, no. 4 (2013): 1125-1129. https://doi.org/10.3892/mmr.2013.1632